You are on page 1of 6

0021-972X/03/$15.

00/0 The Journal of Clinical Endocrinology & Metabolism 88(12):5951–5956


Printed in U.S.A. Copyright © 2003 by The Endocrine Society
doi: 10.1210/jc.2003-031279

Pharmacokinetics and Dose Finding of a Potent


Aromatase Inhibitor, Aromasin (Exemestane), in
Young Males
NELLY MAURAS, JOHN LIMA, DEVAL PATEL, ANNIE RINI, ENRICO DI SALLE, AMBROSE KWOK,
AND BARBARA LIPPE

Nemours Children’s Clinic and Research Programs (N.M., J.L., A.R.), , Jacksonville, Florida 32207; and University of
Florida Health Sciences Center (D.P.) and Amersham Pharmacia Biotech (E.d.S., A.K., B.L.), Peapack, New Jersey 07977

Suppression of estrogen, via estrogen receptor or aromatase 0.002); 50 mg, 32% (P < 0.008)], with a reciprocal increase in
blockade, is being investigated in the treatment of different testosterone concentrations (60% and 56%; P < 0.003 for both).
conditions. Exemestane (Aromasin) is a potent and selective Plasma lipids and IGF-I concentrations were unaffected by
irreversible aromatase inhibitor. To characterize its suppres- treatment. The PK properties of the 25-mg dose showed the
sion of estrogen and its pharmacokinetic (PK) properties in highest exemestane concentrations 1 h after administration,
males, healthy eugonadal subjects (14 –26 yr of age) were re- indicating rapid absorption. The terminal half-life was 8.9 h.
cruited. In a cross-over study, 12 were randomly assigned to Maximal estradiol suppression of 62 ⴞ 14% was observed at
25 and 50 mg exemestane daily, orally, for 10 d with a 14-d 12 h. The drug was well tolerated. In conclusion, exemestane
washout period. Blood was withdrawn before and 24 h after is a potent aromatase inhibitor in men and an alternative to
the last dose of each treatment period. A PK study was per- the choice of available inhibitors. Long-term efficacy and
formed (n ⴝ 10) using a 25-mg dose. Exemestane suppressed safety will need further study. (J Clin Endocrinol Metab 88:
plasma estradiol comparably with either dose [25 mg, 38% (P < 5951–5956, 2003)

T HE BIOLOGICAL ACTIONS of estrogens in males have


begun to be unraveled via prismatic cases of estrogen
deficiency in adults (1–3), gene knockout experiments in
tribute significantly to the changes in body composition and
protein synthesis observed with changing androgen levels in
males. It also suggested that this level of aromatase inhibition
mice (4 – 6), and metabolic studies in vivo (7). Experiments in does not negatively impact markers of bone calcium metab-
both animals and humans, for example, have clearly shown olism, at least in the short term.
that epiphyseal fusion and the completion of final adult A new irreversible aromatase enzyme blocker, exemestane
height are processes regulated by estrogen, even in the male. (Aromasin), offers an alternative to suppress estrogen con-
Male patients with functional mutations in either the estro- centrations. It is structurally related to the natural substrate
gen receptor gene (1) or the aromatase gene (2, 3) have androstenedione, and it is metabolized to an intermediate
demonstrated continued linear growth into adulthood, tall that binds to the active site of the enzyme and inactivates it.
stature, and osteopenia. This information has lead to the It is excreted in the urine and feces. It decreases estradiol
investigation of pharmacologically induced estrogen defi- concentrations in postmenopausal women, but has no effect
ciency as an adjunct in delaying epiphyseal fusion in males on the synthesis of glucocorticosteroids or aldosterone (12,
with short stature and potentially increasing final adult 13). All pharmacokinetic (PK) data available to date are from
height (8, 9).
postmenopausal women, as it is presently used in this pop-
The biosynthesis of estrogens from C19 steroids is regu-
ulation for the treatment of metastatic breast cancer (12–15).
lated by the aromatase cytochrome p450 (CYP19), a product
A daily dose of 25 mg has been shown to have no effect on
of a single CYP19 gene. This enzyme, which catalyzes the
circulating testosterone concentrations in females. This study
conversion of androstenedione and testosterone to estrone
and estradiol, is widely expressed in numerous tissues, in- was designed with two aims: first, to investigate the dose of
cluding bone (10, 11). We have conducted detailed studies of exemestane that can be safely given in adolescent/young
the metabolic effects of selective estrogen suppression in adult males with minimal or no side-effects, and second, to
young eugonadal males using anastrozole, a potent and se- investigate the PK and pharmacodynamics of this aromatase
lective nonsteroidal aromatase inhibitor, and have shown inhibitor in males.
that specific blockade of the aromatase enzyme for 10 wk did
not have catabolic effects on protein metabolism, body com- Subjects and Methods
position, measures of muscle strength, and bone calcium
metabolism (7). The data suggest that estrogens do not con- These studies were approved by the Nemours Children’s Clinic clin-
ical research review committee and Baptist Medical Center/Wolfson
Children’s Hospital institutional review board. Healthy lean male vol-
Abbreviations: AUC, Area under the curve; CBC, cell blood count; unteers between 14 –26 yr of age were recruited after giving informed
HDL, high density lipoprotein; LDL, low density lipoprotein; PK, written consent to participate in study I or II (see below). Their clinical
pharmacokinetic. characteristics are summarized in Table 1.

5951

Downloaded from https://academic.oup.com/jcem/article-abstract/88/12/5951/2661508


by guest
on 28 February 2018
5952 J Clin Endocrinol Metab, December 2003, 88(12):5951–5956 Mauras et al. • Pharmacokinetics of Exemestane in Males

TABLE 1. Clinical characteristics of the study subjects spiked with each of the tritiated hormones, were included in each an-
alytical run to calculate overall recoveries to correct results measured by
Study I Study II RIA. All extractions were performed singly, and all RIA analysis were
(n ⫽ 12) (n ⫽ 10) performed in duplicate. The lower limit of sensitivity was 0.7 pg/ml for
Race, n (%) estradiol, 1.8 pg/ml for estrone, 6 pg/ml for estrone sulfate, 40 pg/ml
White 8 6 for androstenedione, and 30 pg/ml for testosterone. The overall inter-
Black 1 1 assay coefficients of variation were: estradiol, 6.2%; estrone, 12.9%; es-
Hispanic 3 1 trone sulfate, 8.2%; androstenedione, 12.6%; and testosterone, 8.6%. Free
Other 2 testosterone was measured by a validated gas chromatography/mass
Age (yr) spectrometry bioanalytical method at Taylor Technology, Inc. (Prince-
Mean ⫾ SD 18.8 ⫾ 3.5 19.4 ⫾ 3.8 ton, NJ). LH and FSH were measured by RIA at the Nemours Biomedical
Range 14.0 –25.0 14.0 –26.0 Research Laboratory using commercial kits from Diagnostic Systems
BMI (kg/m2) Laboratories, Inc. (Webster, TX). All other hormones were measured at
Mean ⫾ SD 24.0 ⫾ 2.9 25.4 ⫾ 3.0 a contract laboratory by RIAs using commercial kits. All samples were
Range 20.0 –29.0 20.9 –29.1 run in the same assay run. Concentrations of plasma lipids, chemistry
profile, and CBC were measured using automated analyzers at Baptist
Study I: dose finding Medical Center (Jacksonville, FL). Exemestane and 17-hydroexemestane
plasma levels were measured at Pharma Bio Research (Assen, The Neth-
Two different doses of exemestane (Aromasin, 25-mg tablets) were erlands) using a validated liquid chromatography method with tandem
administered orally in random order for 10 d with a 14-d washout in mass spectrometry detection (17). The lower limit of sensitivity was 0.1
between. Twelve subjects were divided into 2 groups (treatment se- ng/ml for both assays.
quences): group I received 25 mg in period 1 and 50 mg in period 2, and
group II received 50 mg in period 1 and 25 mg in period 2. Blood was
withdrawn in the morning, between 0800 – 0900 h at the beginning of Statistical analysis
each treatment cycle and 24 h after the last dose of each treatment cycle
(4 blood draws) for various pharmacodynamic assays. These included For the pharmacodynamic results of study I, descriptive statistics
estradiol, estrone, estrone sulfate, androstenedione, testosterone, free were generated for the assays measured by group (sequence) and by
testosterone, dehydroepiandrosterone sulfate, cortisol, SHBG, IGF-I, treatment. The mean concentrations at baseline and at the end of treat-
IGF-binding protein-3, and plasma lipid profiles [triglycerides, total ment were summarized by period and by treatment group for all assays.
cholesterol, high density lipoprotein (HDL) cholesterol, and low density A paired t test was used to test the difference between baseline and day
lipoprotein (LDL) cholesterol]. Safety data, including general chemis- 10 concentrations for each assay. A cross-over ANOVA with factors for
tries, cell blood count (CBC), urinalysis, and liver profiles, were mea- period, treatment, group (sequence), and subject within group was con-
sured as well. All adverse events were recorded. ducted (18). A baseline value was added to the model. A paired t test
was used to test the difference in concentrations at baseline and on d 10
Study II: PK study for all assays. Significance was established at P ⬍ 0.05.

Ten male volunteers participated in this study arm. They came to the
Clinical Research Center at 0700 h after an overnight fast. An iv heparin PK analysis
lock was placed in a forearm vein for the blood drawing after numbing
the skin with a topical anesthetic (EMLA, AstraZeneca, Wilmington, The PK of exemestane were determined by noncompartmental anal-
DE). In addition to safety laboratories (CBC, chemistry profile, and ysis (19) using the computer program WinNonlin (Pharsight Corp.,
urinalysis), blood was withdrawn for determining exemestane, its me- Mountain View, CA). The maximum plasma concentration was the
tabolite 17-hydroexemestane, estradiol, testosterone, LH, and FSH con- highest concentration observed for each individual. The area under the
centrations. A regular breakfast was served that contained 30% of the curve (AUC) was calculated using the linear trapezoidal rule up to
total calories as fat, and a single dose of 25 mg exemestane was given the last quantifiable concentration and extrapolated to infinite time
with the meal. Blood was withdrawn at 0, 1, 2, 3, 4, 8, 12, 24, 48, 72, 144, (AUC0-inf). The half-life of the terminal decay phase, t1/2,z, was deter-
and 240 h after the administration of exemestane for the same assays as mined by linear regression analysis of the natural log concentration vs.
at baseline. The subjects were fed a regular diet and were free to move time curve, where t1/2,z ⫽ ln2/Kel, where Kel is the slope of the regres-
around. After the 24 h sample was withdrawn, subjects were discharged sion line. Oral clearance was calculated as oral dose/AUC0-inf. Analo-
home, and the 48, 72, 144, and 240 h samples were obtained as out- gous calculations were performed on (c ⫻ t) vs. time plots to estimate
patients. LH and FSH were only measured up to 24 h. the area under the first moment curve (AUMC0-inf). The mean residence
time was calculated as AUMC/AUC PK parameters were summarized
Assays with descriptive statistics.

As exemestane is a steroid, to eliminate any confounding interference


of this compound and its metabolites on the assays of related endoge- Results
nous steroidal hormones (androgens and estrogens), careful separation
of the given compounds in the plasma samples was performed using Study I: dose finding
HPLC, followed by RIA, as described by Johannessen et al. (16). Plasma Analysis of the data on hormone concentrations after the
estradiol, estrone, estrone sulfate, testosterone, and androstenedione
concentrations were measured by a validated HPLC-RIA method at 25- and 50-mg doses showed no difference in any of the
Aster-Cephac Laboratories (Saint-Benoit, Cedex, France). Briefly, a 2-ml parameters measured due to an order effect; hence, the data
plasma sample was loaded onto Amprep C18 cartridge and the fraction were grouped for analysis by dose. The 25- and 50-mg doses
containing estrone sulfate or free steroids (estradiol, estrone, andro- of daily exemestane had comparable effects in suppressing
stenedione, and testosterone) were eluted with 4 ml 24% acetonitrile in
water or 100% acetonitrile, respectively. Estrone sulfate was hydrolyzed
circulating estrogen concentrations, with 38 ⫾ 24% (mean ⫾
with arylsulfatase, and the deconjugated estrone was further extracted sd; P ⫽ 0.002 vs. baseline) and 32 ⫾ 29% (P ⫽ 0.008) decreases
with a C18 cartridge. The extracted fractions were injected into a reverse in estradiol concentrations, 71 ⫾ 12% (P ⬍ 0.0001) and 74 ⫾
phase HPLC system. The eluates corresponding to estradiol, estrone, 12% (P ⬍ 0.0001) decreases in estrone concentrations, and
androstenedione, and testosterone or to deconjugated estrone (to mea-
sure estrone sulfate) were collected and subjected to specific RIAs using
45 ⫾ 27% (P ⫽ 0.004) and 51 ⫾ 20% (P ⫽ 0.02) decreases in
commercial kits. The collected fractions were evaporated and reconsti- estrone sulfate concentrations after doses of 25 and 50 mg,
tuted using an assay buffer before RIA. Appropriate plasma samples, respectively. There was an increase in circulating testoster-

Downloaded from https://academic.oup.com/jcem/article-abstract/88/12/5951/2661508


by guest
on 28 February 2018
Mauras et al. • Pharmacokinetics of Exemestane in Males J Clin Endocrinol Metab, December 2003, 88(12):5951–5956 5953

one concentrations after both 25 mg (60 ⫾ 58%; P ⫽ 0.001) and not the 50-mg dose. Similarly, IGF-binding protein-3 showed
50 mg (56 ⫾ 48%; P ⫽ 0.003) exemestane. Androstenedione a trend toward lower concentrations after the 25-mg dose
concentrations were increased as well after 25 mg (32 ⫾ 36%; (⫺7 ⫾ 13%; P ⫽ 0.09), but not the 50-mg dose. There were no
P ⫽ 0.004) and 50 mg (47 ⫾ 59%; P ⫽ 0.052) exemestane, changes in circulating serum triglycerides, cholesterol, or
respectively (Fig. 1 and Table 2). SHBG concentrations were LDL or HDL cholesterol concentrations with either dose of
decreased by 21 ⫾ 7% (P ⫽ 0.0003) and 19 ⫾ 39% (P ⫽ 0.18) exemestane. Table 2 summarizes the results of the hormonal
at 25 and 50 mg exemestane, respectively. Free testosterone and lipid data.
concentrations were increased by 117 ⫾ 74% (P ⫽ 0.0001) and
154 ⫾ 95% (P ⬍ 0.0001) at both doses, due to the decrease in
Study II: PK
SHBG and the increase in total testosterone. No effect on
circulating dehydroepiandrosterone sulfate was observed at As the level of suppression of circulating estrogens was
either dose. Serum cortisol concentrations increased signif- comparable between doses, we elected to use 25 mg for the
icantly (38 ⫾ 39%; P ⫽ 0.008) with the 25-mg dose, but not subsequent PK study. In all individuals, the highest concen-
the 50-mg dose, yet the increase was well within the normal trations of exemestane were observed in the first blood sam-
range of cortisol concentrations. Plasma IGF-I decreased sig- ple drawn 1 h after oral administration, indicating rapid
nificantly (⫺13 ⫾ 11%; P ⫽ 0.008) after the 25-mg dose, but absorption of the drug. Plasma concentration vs. time profiles

FIG. 1. Estrogen and androgen plasma levels after 10 d of daily exemestane (25 or 50 mg) in healthy young males (mean ⫾ SD; n ⫽ 9 –11). To
convert to Systeme International units: estradiol, picomoles per liter (⫻3.671); estrone, picomoles per liter (⫻3.699); androstenedione, nanomoles
per liter (*0.003492); and testosterone, nanomoles per liter (⫻0.03467).

Downloaded from https://academic.oup.com/jcem/article-abstract/88/12/5951/2661508


by guest
on 28 February 2018
5954 J Clin Endocrinol Metab, December 2003, 88(12):5951–5956 Mauras et al. • Pharmacokinetics of Exemestane in Males

TABLE 2. Changes in hormone and lipid concentrations in study I: young male subjects received 25 or 50 mg exemestane daily for 10
days

End of 10-d % Change from


Dose Baseline P value
Assay n treatment baseline
(mg) (mean ⫾ SD) (end ⫺ baseline)
(mean ⫾ SD) (mean ⫾ SD)
Free testosterone (ng/dl) 25 11 9.5 ⫾ 3.3 19.1 ⫾ 4.7 117.0 ⫾ 73.9 0.0001
50 10 8.2 ⫾ 2.9 19.4 ⫾ 4.5 153.6 ⫾ 94.6 0.0000
DHEAS (ng/ml) 25 11 1561 ⫾ 826 1662 ⫾ 726 18.6 ⫾ 39.9 0.4227
50 9 1771 ⫾ 909 1876 ⫾ 840 2.8 ⫾ 12.5 0.7804
Cortisol (␮g/dl) 25 9 10.2 ⫾ 3.4 13.1 ⫾ 2.7 37.9 ⫾ 39.5 0.0080
50 9 11.8 ⫾ 6.6 11.3 ⫾ 3.5 34.2 ⫾ 104.0 0.7781
SHBG (nmol/liter) 25 10 22 ⫾ 7 18 ⫾ 5 ⫺20.6 ⫾ 7.0 0.0003
50 10 28 ⫾ 20 19 ⫾ 5 ⫺18.9 ⫾ 39.2 0.1756
IGF-I (ng/ml) 25 11 533 ⫾ 137 455 ⫾ 80 ⫺12.5 ⫾ 11.1 0.0075
50 10 491 ⫾ 149 471 ⫾ 118 2.0 ⫾ 19.4 0.8197
IGFBP-3 (ng/liter) 25 11 5.0 ⫾ 0.9 4.6 ⫾ 0.6 ⫺7.0 ⫾ 12.5 0.0878
50 10 4.8 ⫾ 0.5 4.7 ⫾ 0.6 0.2 ⫾ 8.1 0.9776
Triglycerides (mg/dl) 25 11 89.9 ⫾ 57.8 86.2 ⫾ 49.4 ⫺0.8 ⫾ 26.4 0.5821
50 10 118.5 ⫾ 145.1 93.6 ⫾ 51.1 28.0 ⫾ 60.3 0.5634
Cholesterol (mg/dl) 25 11 144 ⫾ 11 142 ⫾ 17 ⫺1.3 ⫾ 9.3 0.6513
50 10 139 ⫾ 15 145 ⫾ 14 4.2 ⫾ 6.3 0.0725
Cholesterol HDL (mg/dl) 25 11 42 ⫾ 11 42 ⫾ 12 ⫺1.0 ⫾ 7.4 0.6938
50 10 43 ⫾ 11 41 ⫾ 11 ⫺4.1 ⫾ 13.0 0.2796
Cholesterol LDL (mg/dl) 25 11 107 ⫾ 16 106 ⫾ 15 0.4 ⫾ 15.6 0.8423
50 10 96 ⫾ 25 108 ⫾ 13 20.7 ⫾ 48.1 0.1381
DHEAS, Dehydroepiandrosterone sulfate; IGFBP-3, IGF-binding protein-3. To convert to Systeme International units: free testosterone,
nmol/liter (⫻0.03467); DHEAS, ␮mol/liter (⫻0.002714); cortisol, nmol/liter (⫻27.59); IGF-I, mg/liter (⫻1); triglycerides, mmol/liter (⫻0.01129);
cholesterol, mmol/liter (⫻0.02586).

FIG. 2. Mean ⫾ SD exemestane plasma concen-


trations vs. time in 10 young males receiving a
single 25-mg oral dose.

in all subjects were characterized by a biexponential decline estradiol suppression, plasma testosterone levels were un-
in exemestane (Fig. 2), with terminal half-life of 8.9 h. The changed and thereafter tended to increase by 32% between
other PK parameters are listed in Table 3. The mean maximal 2–3 d; however, contrary to the significant increase in tes-
plasma concentration of the metabolite 17-hydroexemestane tosterone observed after 10-d daily dosing, this change did
was 1.16 ⫾ 0.36 ng/ml, a concentration achieved 1 h after the not achieve statistical significance after a single oral dose.
exemestane dose. These levels rapidly declined, and con- Serum LH and FSH concentrations were measured up to 24 h
centrations below the lower limit of sensitivity (0.1 ng/ml) at the same time intervals as the exemestane samples for the
were observed at a median time of 12 h (range, 4 –24 h). PK analysis. The mean baseline levels of LH and FSH were
The mean baseline levels of estradiol and testosterone 4.8 ⫾ 2.2 and 1.3 ⫾ 0.7 mIU/ml, respectively. The percent
were 24.5 ⫾ 8.8 pg/ml and 581 ⫾ 165 ng/dl, respectively. change from baseline up to 24 h is reported in Fig. 4. The LH
Maximal suppression of estradiol (62 ⫾ 14%) was observed levels initially decreased by 26% at 2 h; thereafter, there was
12 h after a single 25-mg dose of exemestane. Estradiol re- a tendency for an increase to a maximum of 81% at 24 h. The
mained suppressed by 58 ⫾ 21% at 24 h and returned to levels of FSH were unchanged up to 12 h and increased by
baseline 3– 6 d after treatment (Fig. 3). At the time of maximal 49% at 24 h.

Downloaded from https://academic.oup.com/jcem/article-abstract/88/12/5951/2661508


by guest
on 28 February 2018
Mauras et al. • Pharmacokinetics of Exemestane in Males J Clin Endocrinol Metab, December 2003, 88(12):5951–5956 5955

TABLE 3. Pharmacokinetic parameters after a single 25-mg oral


dose of exemestane in 10 young males

PK parameter Mean ⫾ SE

Cmax (ng/ml) 16.1 ⫾ 8.0


AUC (ng/ml䡠h) 36.4 ⫾ 8.8
Kel (h⫺1) 0.0955 ⫾ 0.0363
Half-life (h) 8.9 ⫾ 5.3
MRT (h) 7.0 ⫾ 3.7
Oral clearance (liter/h) 725 ⫾ 186
Cmax, Maximum plasma concentration; Kel, slope of the regres-
sion line; MRT, mean residence time.

FIG. 4. Percent change from baseline (mean ⫾ SD) in plasma LH and


FSH concentrations after a single 25-mg dose of exemestane in 10
young males.

to a true gender dependency possibly involving the volume


of distribution (lower in males than females) and plasma or
tissue protein binding (respectively, higher and lower in
males). This finding may also be due to the lower sensitivity
of the analytical methodology used in the previous studies
(14 pg/ml by HPLC/RIA) (21).
The maximal suppression evoked by exemestane at the
single dose of 25 mg in the present study was similar to
published results in postmenopausal women, but the time
course differed (24). Evans et al. (24) reported that a single
25-mg oral dose of exemestane maximally suppressed estra-
FIG. 3. Percent change from baseline (mean ⫾ SD) in plasma estradiol
concentrations after a single 25-mg dose of exemestane in 10 young
diol concentrations by 72% 3 d after administration, and
males. estradiol levels returned to baseline only 8 –11 d after drug
administration. In the present study maximal suppression of
Safety estradiol of 62% was observed 12 h after exemestane admin-
istration and returned to baseline 3– 6 d after administration.
Exemestane was well tolerated by the study subjects, with The reason for this difference is not clear, but may be related
no serious adverse events reported. General chemistries, to the shorter half-life of exemestane in males, the lower
CBC, and differential urinalysis and liver profiles were mea- exposure to exemestane, and the higher levels of the aro-
sured and were unchanged during administration. matase substrates androstenedione (⬃1 ng/ml in young
males vs. ⬃0.5 ng/ml in postmenopausal women), particu-
Discussion larly the much higher testosterone concentrations in young
We report the first detailed study of the pharmacological males than in postmenopausal women (⬃700 ng/dl vs. ⬃20
effects of exemestane in male subjects. Doses of 25 and 50 mg ng/dl, respectively) (25). This is supported by the observa-
were comparable in suppressing all circulating estrogens and tion that in the 10-d study in young males reported here, the
had similar effects of increasing serum androstenedione and suppression of estradiol is weaker (due to the very high
testosterone concentrations. There were 38%, 71%, and 45% levels of the precursor testosterone) than that of estrone (due
decreases in estradiol, estrone, and estrone sulfate concen- to androstenedione levels not very different from those in
trations, respectively, after 10 d, approximately 24 h after postmenopausal women). A limited suppression of circulat-
administration of the last dose of 25 mg exemestane, coupled ing estradiol (⬃50%) has been reported in a similar study in
with 60% and 32% increases in testosterone and andro- young males treated with 1 mg daily anastrozole (7), a dose
stenedione concentrations. The rise in the aromatase sub- that reduces estradiol by 85% in postmenopausal women
strates, testosterone and androstenedione, is probably sec- (23).
ondary to substrate accumulation and/or to the feedback Aromatase inhibitors are being investigated in a variety of
increase in gonadotropins caused by aromatase blockade. clinical situations besides breast cancer. As estrogen is the
The 21% decrease in SHBG concentrations caused by 25 mg principal factor responsible for epiphyseal fusion, aromatase
exemestane confirms the observation in postmenopausal blockers are being studied in the treatment of severe short
women (20). stature in boys (8, 9). This class of compounds has a theo-
The maximum plasma concentration, time to achieve max- retical advantage over using LHRH analogs to delay puberty,
imal concentrations and oral clearance for exemestane after because they allow for progressive virilization while de-
oral administration of a single dose of 25 mg in the present creasing estrogens, potentially extending the time of epiph-
study of males were similar to those reported for females yseal fusion and thus the time for linear growth. Trials have
(21–23). The terminal half-life in the present study (8.9 h) was not yet been performed in adolescent females due to the
considerably shorter than the published value of 27 h (23). concerns that increased circulating gonadotropins, de-
The reason for this difference is not clear, but may be related creased estrogens, and increased testosterone could precip-

Downloaded from https://academic.oup.com/jcem/article-abstract/88/12/5951/2661508


by guest
on 28 February 2018
5956 J Clin Endocrinol Metab, December 2003, 88(12):5951–5956 Mauras et al. • Pharmacokinetics of Exemestane in Males

itate ovarian dysfunction and virilization, as is seen in the (GHD) boys in puberty: preliminary results [Abstract OR-9-99]. LWPES/ESPE
6th Joint Meeting of the Society for Pediatr Research, Montréal, Canada, 2001
aromatase-deficient female (3, 26). Because of its properties 10. Simpson ER, Zhao Y, Agarwal VR, Michael MD, Bulun SE, Hinshelwood
of increasing circulating gonadotropins, it has been used as MM, Graham-Lorence S, Sun T, Fisher CR, Qin K, Mendelson CR 1997
a treatment for oligospermic men with low testosterone/ Aromatase expression in health and disease. Recent Prog Horm Res 52:185–
214
estradiol ratios with initial preliminary success (27). This 11. Sasano H, Uzuki M, Sawai T, Nagura H, Matsunaga G, Kashimoto O, Harada
class of compounds also has a theoretical application in the N 1997 Aromatase in human bone tissue. J Bone Miner Res 12:1416 –1423
treatment of gynecomastia in those individuals with over- 12. Clemett D, Lamb HM 2000 Exemestane: a review of its use in postmenopausal
women with advanced breast cancer. Drugs 59:1279 –1296
expressed aromatase activity as recently reported (28). In 13. 2003 Aromasin. In: Physicians desk reference, 57th ed. Montvale, NJ: Thomson
addition, studies conducted using estrogen receptor block- PDR; 2692–2695
14. Lonning PE, Bajetta E, Murray R, Tubiana-Hulin M, Eisenberg PD, Mick-
ade in the treatment of gonadotropin-independent preco- iewicz E, Celio L, Pitt P, Mita M, Aaronson NK, Fowst C, Arkhipov A, di Salle
cious puberty have shown encouraging results (29); hence, E, Polli A, Massimini G 2000 Activity of exemestane in metastatic breast
the use of aromatase blockers seems like a natural alternative cancer after failure of non steroidal aromatase inhibitors: a phase II trial. J Clin
Oncol 18:2234 –2244
worthy of clinical trials as well. 15. Kaufmann M, Bajetta E, Dirix LY, Fein LE, Jones SE, Zilembo N, Dugardyn
We conclude that exemestane is a potent aromatase in- JL, Nasurdi C, Mennel RG, Cervek J, Fowst C, Polli A, di Salle E, Arkhipov
hibitor in men. Exemestane appears to be an alternative in the A, Piscitelli G, Miller LL, Massimini G 2000 Exemestane is superior to
megestrol acetate after tamoxifen failure in postmenopausal women with
choice of inhibitors of the aromatase enzyme available for advanced breast cancer: results of a phase III randomized double-blind trial.
human studies. Further studies are underway to estimate J Clin Oncol 18:1399 –1411
16. Johannessen DC, Engan T, Di Salle E, Zurlo MG, Paolini J, Ornati G,
dose and dosing intervals that will provide therapeutic sup- Piscitelli G, Kvinnsland S, Lonning PE 1997 Endocrine and clinical effects of
pression of estrogen concentrations in males. Long-term exemestane (PNU 155971), a novel steroidal aromatase inhibitor, in postmeno-
safety will also require further investigation. pausal breast cancer patients: a phase I study. Clin Cancer Res 3:1101–1108
17. Cenacchi V, Barattč S, Cicioni P, Frigerio E, Long J, James C 2000 LC-MS-MS
determination of exemestane in human plasma with heated nebulizer interface
Acknowledgments following solid-phase extraction in the 96 well plate format. J Pharmacol Biom
Anal 22:451– 460
We are grateful to Burnese Rutledge and the nursing staff of Wolfson 18. Hills M, Armitage P 1979 The two-period cross-over clinical trial. Br J Clin
Children’s Hospital Clinical Research Center, to Brenda Sager and the Pharmacol 8:7–20
Biomedical Analysis Laboratory, and to Steve Fordham and Holly 19. Gibaldi M, Perrier D 1982 Pharmacokinetics, 2nd Ed. New York: Marcel
Murphy. Dekker
20. Zilembo N, Noberasco C, Bajetta E, Martinetti A, Mariani L, Orefice S,
Buzzoni R, Di Bartolomeo M, Di Leo A, Laffranchi A 1995 Endocrinological
Received July 23, 2003. Accepted September 10, 2003. and clinical evaluation of exemestane, a new steroidal aromatase inhibitor. Br J
Address all correspondence and requests for reprints to: Nelly Mau- Cancer 72:1007–1012
ras, M.D., Nemours Children’s Clinic, 807 Children’s Way, Jacksonville, 21. Poggesi I, Jannuzzo MG, di Salle E, Piscitelli G, Rocchetti M, Spinelli R,
Florida 32207. E-mail: nmauras@nemours.org. Broutin F, Ornati G, Massimini G 1999 Effect of food and formulation on the
This work was supported by a grant from Amersham Pharmacia pharmacokinetics (PK) and pharmacodynamics (PD) of a single oral dose of
exemestane (Aromasin®, EXE) [Abstract 741]. Proc Am Soc Clin Oncol 18:193a
Biotech. 22. Spinelli R, Jannuzzo MG, Poggesi I 1999 Pharmacokinetics (PK) of Aromasin
(Exemestane, EXE) after single and repeated doses in healthy postmenopausal
References volunteers (HPV) [Abstract 1185]. Eur J Cancer 35:S295
23. Buzdar AU, Robertson JFR, Eiermann W, Nabholtz JM 2002 An overview of
1. Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, Specker B, Williams the pharmacology and pharmacokinetics of the newer generation aromatase
TC, Lybahn DB, Korach, KS 1994 Estrogen resistance caused by a mutation inhibitors anastrozole, letrozole and exemestane. Cancer 95:2006 –2016
in the estrogen-receptor gene in a man. N Engl J Med 331:1056 –1061 24. Evans TR, Di Salle E, Ornati G, Lassus M, Benedetti MS, Pianezzola E,
2. Carani C, Qin K, Simoni M, Faustini-Fustini M, Serpente S, Boyd J, Korach Coombes RC 1992 Phase I and endocrine study of exemestane (FCE 24304), a
KS, Simpson ER 1997 Effect of testosterone and estradiol in man with aro- new aromatase inhibitor in postmenopausal women. Cancer Res 52:5933–5939
matase deficiency. N Engl J Med 337:95 25. Kvinnsland S, Anker G, Dirix LY, Bonneterre J, Prove AM, Wilking N,
3. Morishima A, Grumbach MM, Simpson ER, Fisher C, Qin K 1995 Aromatase Lobelle JP, Mariani O, di Salle E, Polli A, Massimini G 2000 High activity
deficiency in male and female siblings caused by a novel mutation and the and tolerability demonstrated for exemestane in postmenopausal women with
physiological role of estrogens. J Clin Endocrinol Metab 80:3689 –3698 metastatic breast cancer who had previously failed on tamoxifen treatment.
4. Simpson ER 1998 Genetic mutations resulting in estrogen insufficiency in the Eur J Cancer 36:976 –982
male. Mol Cell Endocrinol 145:55–59 26. Mullis PE, Yoshimura N, Kuhlmann B, Lippuner K, Jaeger P, Harada H 1997
5. Fisher CR, Graves KH, Parlow AF, Simpson ER 1998 Characterization of mice Aromatase deficiency in a female who is compound heterozygote for two new
deficient in aromatase (ArKO) because of targeted disruption of the cyp19 point mutations in the P450arom gene: impact of estrogens on hypergonado-
gene. Proc Natl Acad Sci USA 95:6965– 6970 tropic hypogonadism, multicystic ovaries, and bone densitometry in child-
6. Hewitt SC, Korach KS 2002 Estrogen receptors: structure, mechanisms and hood. J Clin Endocrinol Metab 82:1739 –1745
function. Rev Endocr Metab Disord 3193–3200 27. Raman JD, Schlegel PN 2002 Aromatase inhibitors for male infertility. J Urol
7. Mauras N, O’Brien KO, Oerter Klein K, Hayes V 2000 Estrogen suppression 167:624 – 629
in males: metabolic effects. J Clin Endocrinol Metab 85:2370 –2377 28. Shozu M, Sebastian S, Takayama K, Hsu WT, Schultz RA, Neely K, Bryant
8. Wickman S, Sipila I, Ankarberg-Lindgren C, Norjavaara E, Dunkel L 2001 M, Bulun SE 2003 Estrogen excess associated with novel gain-of-function
A specific aromatase inhibitor and potential increase in adult height in boys mutations affecting the aromatase gene. N Engl J Med 348:1855–1865
with delayed puberty: a randomized controlled trial. Lancet 357:1743–1748 29. Eugster E, Rubin S, Reiter E, Plourde P, Jou HC, Pescovitz O 2003 Tamoxifen
9. Mauras N, Rini A, Welch S, Oerter-Klein K, Long term effects of combined treatment for precocious puberty (PP) in McCune-Albright syndrome: mul-
treatment with an aromatase blocker (Arimidex®) and GH in GH deficient ticenter trial. J Pediatr 143:60 – 66

Downloaded from https://academic.oup.com/jcem/article-abstract/88/12/5951/2661508


by guest
on 28 February 2018

You might also like